Alma Mater Studiorum Università di Bologna · Alma Mater Studiorum – Università di Bologna...

148
Alma Mater Studiorum Università di Bologna DOTTORATO DI RICERCA IN Oncologia e Patologia Sperimentale Ciclo XXVII° Settore Concorsuale di afferenza: 06/A2 Settore Scientifico disciplinare: MED/04 Cancer and aging: a multidisciplinary medicinal chemistry approach on relevant biological targets such as proteasome, sirtuins and interleukin 6 Presentata da: Marco Daniele Parenti Coordinatore Dottorato Relatore Prof. Pier Luigi Lollini Prof. Stefano Salvioli Esame finale anno 2015

Transcript of Alma Mater Studiorum Università di Bologna · Alma Mater Studiorum – Università di Bologna...

  • Alma Mater Studiorum – Università di Bologna

    DOTTORATO DI RICERCA IN

    Oncologia e Patologia Sperimentale

    Ciclo XXVII°

    Settore Concorsuale di afferenza: 06/A2

    Settore Scientifico disciplinare: MED/04

    Cancer and aging: a multidisciplinary medicinal

    chemistry approach on relevant biological targets

    such as proteasome, sirtuins and interleukin 6

    Presentata da: Marco Daniele Parenti

    Coordinatore Dottorato Relatore

    Prof. Pier Luigi Lollini Prof. Stefano Salvioli

    Esame finale anno 2015

  • 2

    Table of Content

    Preface ............................................................................................................................................ 5

    List of Abbreviations ...................................................................................................................... 7

    1. Introduction ............................................................................................................................. 9

    1.1 The Drug Discovery Process ............................................................................................. 9

    1.2 Molecular Modeling and rational drug design in drug discovery ....................................... 9

    1.2.1 Ligand based approaches ......................................................................................... 11

    1.2.2 Structure based approach ......................................................................................... 12

    1.3 Cancer and Aging in drug discovery ............................................................................... 12

    1.4 Selected Targets and their Involvement in Human Pathologies ........................................ 13

    2. Aim of the Thesis .................................................................................................................. 15

    3. Proteasome and Immunoproteasome Inhibitors ...................................................................... 16

    3.1 Proteasomes: mandatory terminators. .............................................................................. 16

    3.2 Why is i-proteasome a potential therapeutic target? ......................................................... 18

    3.3 I-proteasome as target for cancer therapy. ....................................................................... 19

    3.3.1 I-proteasome in Multiple Myeloma .......................................................................... 20

    3.3.2 I-proteasome in solid tumours. ................................................................................. 21

    3.3.3 I-proteasome as a target for immunotherapy? ........................................................... 26

    3.4 I-proteasome as target for neuropathologies .................................................................... 26

    3.4.1 Alzheimer disease. ................................................................................................... 27

    3.4.2 Multiple Sclerosis. ................................................................................................... 28

    3.4.3 Temporal Lobe Epilepsy. ......................................................................................... 29

    3.5 An overview of selective s- and i-proteasome inhibitors and enhancers. .......................... 30

    3.6 Human immunoproteasome model. ................................................................................. 33

    3.7 Computer-aided drug design approaches ......................................................................... 38

  • 3

    3.8 Hit Identification ............................................................................................................. 39

    3.9 Conclusion and perspective ............................................................................................. 46

    3.10 Experimental Section................................................................................................... 47

    4. Sirtuins in drug discovery ...................................................................................................... 48

    4.1 Sirtuins: at the crossroad of metabolism, cancer, and inflammation ................................. 48

    4.2 Sirtuins and cancer .......................................................................................................... 48

    4.3 Sirtuin inhibitors ............................................................................................................. 50

    4.4 Sirtuins activators ........................................................................................................... 52

    5. Development of a Sirtuins selectivity model .......................................................................... 55

    5.1 Sequence and structural comparison of the catalytic cores ............................................... 57

    5.2 Structural comparison of the binding sites ....................................................................... 60

    5.3 Structural superposition of available three-dimensional structures ................................... 61

    5.4 Conclusions .................................................................................................................... 70

    5.5 Experimental Procedures................................................................................................. 71

    6. Discovery of new SIRT3 modulators ..................................................................................... 73

    6.1 Targeting SIRT3 with structure-based drug design techniques ........................................ 74

    6.2 Evaluation of acetylation pattern of mitochondrial proteins ............................................. 76

    6.2.1 Evaluation of acetylation pattern of a specific protein target of SIRT3 ..................... 77

    6.3 Evaluation of cell response to toxic stimulation ............................................................... 78

    6.4 Conclusions .................................................................................................................... 79

    6.5 Experimental Section ...................................................................................................... 79

    7. Discovery of new SIRT6 inhibitors ....................................................................................... 81

    7.1 Structure-based in silico screening .................................................................................. 82

    7.2 Selectivity profiling ........................................................................................................ 85

    7.3 Biological characterization of the identified SIRT6 inhibitors ......................................... 89

    7.4 Conclusions .................................................................................................................... 92

  • 4

    7.5 Experimental section ....................................................................................................... 93

    8. Optimization of SIRT6 inhibitors - Quinazolinedione derivatives .......................................... 97

    8.1 Selection of analog candidates for biological testing ....................................................... 97

    8.2 Selectivity profiling ...................................................................................................... 102

    8.3 Mechanism for SIRT6 inhibition by quinazolinediones ................................................. 103

    8.4 Biological evaluation of quinazolinedione SIRT6 inhibitors .......................................... 104

    8.5 Quinazolinedione inhibitors sensitize cancer cells to chemotherapeutics ....................... 107

    8.6 Conclusions .................................................................................................................. 110

    8.7 Experimental section ..................................................................................................... 111

    9. Optimization of SIRT6 inhibitors – Salicylate derivatives ................................................... 116

    10. Interleukin 6 inhibitors ..................................................................................................... 118

    10.1 Biologic Functions of IL-6 ........................................................................................ 118

    10.2 IL-6 Signaling Pathway ............................................................................................. 119

    10.3 IL-6 in aging and aging-related diseases .................................................................... 120

    10.4 IL-6 and cancer ......................................................................................................... 121

    10.5 Pharmacological approaches to blockade of IL-6 signaling ........................................ 121

    10.6 Drug repurposing....................................................................................................... 122

    10.7 Screening for IL6 – IL6 receptor interaction modulators ............................................ 122

    10.7.1 Screening of hexameric assembly IL-6/IL-6R/gp130 ligands .................................... 122

    10.7.2 Screening of Protein-Protein Interface inhibitors ....................................................... 126

    10.8 Conclusion and perspective ....................................................................................... 128

    10.9 Experimental section ................................................................................................. 131

    11. Concluding remarks and future perspectives ..................................................................... 132

    Acknowledgements ..................................................................................................................... 133

    References .................................................................................................................................. 134

  • 5

    Preface

    Publications and Patents

    This thesis is based on the following publications and manuscripts:

    Publications:

    1. Parenti MD, Bruzzone S, Nencioni A, Del Rio A. Selectivity hot-spots of sirtuin catalytic

    cores. To be Submitted to Structure

    2. Sociali G, Galeno L, Parenti MD, Grozio A, Bauer I, Passalacqua M, Boero S, Donadini A,

    Millo E, Bellotti M, Sturla L, Franceschi C, Ballestrero A, Poggi A, Bruzzone S, Nencioni

    A, Del Rio A. Quinazolinedione SIRT6 inhibitors sensitize cancer cells to

    chemotherapeutics. To be Submitted to ACS Chemical Biology

    3. Parenti MD, Grozio A, Bauer I, Galeno L, Damonte P, Millo E, Sociali G, Franceschi C,

    Ballestrero A, Bruzzone S, Del Rio A, Nencioni A. Discovery of novel and selective SIRT6

    inhibitors. J Med Chem. 2014 Jun 12;57(11):4796-804.

    4. Bellavista E, Andreoli F, Parenti MD, Martucci M, Santoro A, Salvioli S, Capri M, Baruzzi

    A, Del Rio A, Franceschi C, Mishto M. Immunoproteasome in cancer and neuropathologies:

    a new therapeutic target? Curr Pharm Des. 2013;19(4):702-18.

    5. Bruzzone S, Parenti MD, Grozio A, Ballestrero A, Bauer I, Del Rio A, Nencioni A.

    Rejuvenating sirtuins: the rise of a new family of cancer drug targets. Curr Pharm Des.

    2013;19(4):614-23.

    6. Andreoli F, Barbosa AJ, Parenti MD, Del Rio A. Modulation of epigenetic targets for

    anticancer therapy: clinicopathological relevance, structural data and drug discovery

    perspectives. Curr Pharm Des. 2013;19(4):578-613.

    Patents:

    WO/2014/170875. Quinazolinedione compounds with a sirtuin inhibiting activity.

    Applicants: University of Genoa and Alma Mater Studiorum – University of Bologna.

    Inventors: DEL RIO, Alberto; (IT), FRANCESCHI, Claudio; (IT), PARENTI, Marco,

    Daniele; (IT), BAUER, Inga; (IT), BRUZZONE, Santina; (IT), GROZIO, Alessia; (IT),

    NENCIONI, Alessio; (IT). Priority data: MI2013A000646 19.04.2013 IT.

  • 6

    WO/2014/170873. Compounds with a sirtuin inhibiting activity. Applicants: University of

    Genoa and Alma Mater Studiorum – University of Bologna. Inventors: DEL RIO, Alberto;

    (IT), FRANCESCHI, Claudio; (IT), PARENTI, Marco, Daniele; (IT), BAUER, Inga; (IT),

    BRUZZONE, Santina; (IT), GROZIO, Alessia; (IT), NENCIONI, Alessio; (IT). Priority

    data: MI2013A000647 19.04.2013 IT.

    Author’s contribution

    The rational design and optimization of active molecules through molecular modeling techniques

    was carried out by the candidate in the framework of the BioChemoInformatics Lab (Dept. of

    Experimental, Diagnostic and Specialty Medicine, University of Bologna) while the biological

    testing of compounds was conducted in collaboration with external laboratories.

    Intellectual Property Issues

    Chemical structures of sirtuin 3 modulators discussed in chapter 6 are considered for patenting,

    therefore are not disclosed.

    Salycilate derivatives of sirtuin 6 inhibitors discussed in chapter 8 are currently under biological

    testing, and will be disclosed in a forthcoming publication.

  • 7

    List of Abbreviations

    2-NBDG = 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxy-D-glucose

    AD = Alzheimer‟s disease

    AML = acute myeloid leukemia

    APM = antigen-processing machinery

    APP = amyloid precursor protein

    C-like = caspase-like

    CNS = central nervous system

    CTL = CD8+ citotoxic T cell

    CT-like = chymotrypsin-like

    DUBs = deubiquitylating enzymes

    EAE = autoimmune encephalomyelitis

    ER = endoplasmic reticulum

    ETC = electron transport chain

    HD = Huntington‟s disease

    HMGB1 = high-mobility group box-1

    HTS = High troughput screening

    IL = interleukin

    IL-6 = Interleukin 6

    IL-6R = Interleukin 6 receptor

    ILR1 = interleukin receptor 1

    i-proteasome = immunoproteasome

    IS = immune system

    MBP = myelin basic protein

    MLR = mixed leukocyte reaction

    MM = multiple myeloma

    MS = multiple sclerosis

    NAM = Nicotinamide

    PBMC = peripheral blood mononuclear cells

    PDB = Protein Data Bank

    PHA = phytohemagglutinin

    PHFs = tau-based paired helical filaments

  • 8

    QSAR = quantitative structure-activity relationships

    RMSD = root mean square deviation

    SAR = Structure-Activity relationship

    SBDD = structure-based drug design

    s-proteasome = standard proteasome

    STAC = sirtuin activating compounds

    TcR = T cell receptor

    TLE = temporal lobe epilepsies

    T-like = trypsin-like

    TLR-4 = Toll-like receptor-4

    t-proteasome = thymus proteasome

    T-reg = regulatory T cells

    UPS = Ubiquitin Proteasome System

  • 9

    1. Introduction

    1.1 The Drug Discovery Process

    Drug discovery is an extremely laborious and expensive process, requires an average of 13 years of

    research and an investment of US$1.8 billion to bring a single drug from the bench to a patient's

    bedside1; this is not in the least surprising, when one considers the complexity of biological

    systems, most of which is only now beginning to be understood. Despite increase in investment in

    drug discovery, the output is considerably low due to high rate of drug failure in clinical trials2.

    Consequently, in order to reduce the cost and time of a drug to reach market, new technologies were

    ventured. The genomics and the post-genomics eras, with the parallel advances in high-throughput

    experimental methods and screening techniques to analyze whole genomes and proteomes, are

    witnessing an explosion in the types and amount of information available, not only with respect to

    the genome sequences and protein structures but also with respect to gene-expression, regulation

    and protein–protein interactions. The availability of such information in publicly accessible

    databases and the advances in both computing power as well as in computational methods for data

    mining and modeling, have led to the emergence of several in silico approaches to systematically

    address several questions in biology, with an obvious impact on drug discovery3,4

    . Today, drug

    discovery usually follows the general scheme presented in Figure 1.1. In short, the goal is to

    identify a compound that can modulate the effect of a molecular target that regulates a biological

    process related to a disease. Once a target has been identified and shown to be relevant in a disease

    model, high throughput screening, or its theoretical counterpart, virtual screening, is usually

    employed to generate a set of hit compounds. Following this, some of the promising molecules that

    show good physico-chemical properties are subject to further chemical exploration. The synthesized

    compounds in these lead series are evaluated and a structure–activity relationship (SAR) is derived,

    as well as the pharmacokinetic and pharmacodynamic profiles of the most promising compounds.

    The final phase in preclinical research is the transformation of a lead structure into a candidate drug,

    which is then considered for testing in clinical trials.

    1.2 Molecular Modeling and rational drug design in drug discovery

    Historically, serendipity and trial and error have played a major role in the discovery of drugs. The

    source of active substances has often been medical plants and herbs. With the advent of synthetic

    organic chemistry and modern pharmacology a more systematic search for new pharmaceutically

  • 10

    active compounds began, and these were often evaluated using animal experiments. The chemical

    modification of lead compounds, on a trial-and-error basis, typically led to compounds with

    improved potency, selectivity and bioavailability and reduced toxicity. However, this approach is

    costly and labor- and time-intensive and researchers in the pharmaceutical industry are constantly

    developing methods with a view to increase the efficiency of the drug discovery process. Two

    directions have evolved from these efforts. The „random‟ approach involves the development of

    HTS assays and the testing of a large number of compounds, and combinatorial chemistry is used to

    satisfy the need for extensive compound libraries. The „rational‟, approach relies on the knowledge

    of the structure of the target protein or knowledge about available potential compounds. Rational

    design approach involves the prediction of hypothetical ligands for the target protein from

    molecular modeling and the subsequent chemical synthesis and biological testing of specific

    compounds. Many rational design approaches have been suggested to increase the cost-

    effectiveness of discovery programs. Such approaches include ligand based approaches such as

    pharmacophore modeling, determination of quantitative structure-activity relationships (QSAR),

    which use accumulated information for ligands of previously executed discovery programs, and

    receptor based approaches such as docking, which use available information about target protein

    structure. One of most commonly used rational design approach is the so-called virtual or in silico

    screening; this methodology involves the computational filtering of a large body of molecules (e.g.,

    those comprising a corporate database or a database of commercially available molecules) to

    identify those that have a high probability of activity in the biological test system of interest. Thus a

    virtual screening method takes as input all those molecules that might be acquired (or synthesized)

    and tested, and then outputs those few that should be tested. Although there are numerous methods

    for performing a virtual screening, they can be roughly classified into two main types: ligand-based

    approaches which do not utilize the structure of the biological target in screening, and structure-

    based approaches, which utilize the structure of the biological target, usually obtained by NMR or

    X-ray methods, and a variety of molecular docking algorithms and scoring functions. Hybrid

    approaches which combine aspects from ligand-based and structure-based methods are also

    frequently employed in virtual screening studies.

  • 11

    Figure 1.1 Drug Discovery Pipeline.

    1.2.1 Ligand based approaches

    Ligand-based approaches typically utilize knowledge of a set of compounds with known activity

    against the biological target. These approaches are frequently employed in the absence of structural

    information on the target in question. The key concept in ligand based approaches is that

    compounds that are structurally similar or have similar structural components to the known active

    compounds are more likely to have activity themselves. A variety of ligand-based screening

    methods have been developed, such as substructure and similarity searching, pharmacophore

    searching, clustering methods, and QSAR methods. Among these, the pharmacophore screening is

    certainly one of the most used methods.

    Pharmacophore modeling allows determining the spatial arrangement of chemical features that

    confer drug activity toward a target receptor. Having established the chemical space occupied by

    active ligands, pharmacophore modeling software allows researchers to create 3D structure-activity

    relationships, screen databases, and generate hits without the benefit of a receptor structure.

    Compared to other ligand-based methods provides several advantages: i) can take into account 3D

    conformational variations and functional group properties such as polarity, hydrogen bond

    potential, aromaticity, and hydrophobicity; ii) can identify lead compounds that are structurally

    dissimilar to those already known, a process known as „scaffold-hopping‟ or „lead-hopping‟; iii) can

    also incorporate known structural information of the biological target‟s active site, if any is known,

  • 12

    in the form of exclusion spheres or a molecular surface, both of which create barriers the

    compounds being searched are not permitted to encroach.

    1.2.2 Structure based approach

    In structure-based drug design (SBDD), the knowledge of the three-dimensional structure of a target

    is exploited to design small molecules able to tightly bind the active site and modulate the

    biological function in a desired way; the same tools could be used to assess the selectivity of the

    potential ligands towards different targets, making it easier to develop drugs with fewer side effects.

    This could reduce the time and resources needed to identify new interesting lead compounds. The

    structure-based methods, such as docking, have been successfully used to identify new hits from

    large libraries of chemical compounds and to predict their binding modes and affinities, and

    currently represent one of the primary methodologies used to discover new “hit” compounds.

    Molecular docking involves two main processes: pose prediction and scoring. In pose prediction a

    search algorithm determines an optimal conformation and orientation for a given compound in the

    receptor, or active site. This is followed by scoring to determine whether the pose will be accepted

    or rejected. Therefore, docking techniques enable both to predict the binding mode of a ligand and

    to roughly estimate its binding affinity for the biological target. Several scoring functions have been

    developed to approximate the interaction energy between proteins and related ligands, and all of

    these are simple linear mathematical models that estimate chemical properties (such as shape,

    charge distribution, hydrophobic/hydrophilic potentials and so on) of the molecules.

    1.3 Cancer and Aging in drug discovery

    The ultimate goal of biomedical research is to translate laboratory discoveries or clinical

    observations into new therapies to ameliorate disease and extend life expectancy and quality,

    namely the average total number of years of remaining meaningful life at a given age5. Ageing is a

    complex and multifactorial process characterized by the many forms of damage accumulation at the

    molecular, cellular, and tissue level that progress with advancing age, decreasing the body‟s normal

    response and function. No single theory currently exists that can explain all of the hallmarks of

    ageing, suggesting that ageing is a multi-step and multi-event process6. At first glance, cancer and

    ageing have an inverse relationship because cancer cells are capable of uncontrolled growth and

    division, whereas ageing cells have a diminished proliferative capacity. Indeed, it has been well-

    established that older adults have a higher risk for cancer, as well as a higher risk of the onset of

    others chronic inflammation-associated diseases such as diabetes, stroke, and neurodegenerative

  • 13

    disease. Ageing is involved in a number of events responsible for carcinogenesis and cancer

    development at the molecular, cellular, and tissue levels. Actually, ageing and cancer have common

    origins due to internal and environmental stress and share some common hallmarks such as

    genomic instability, epigenetic alteration, aberrant telomeres, inflammation and immune injury,

    reprogrammed metabolism, and impaired degradation of intracellular biomolecules and organelles7.

    The pharmaceutical industry is always searching for new biological targets to generate novel

    therapies; aging could represent a “blockbuster” market because the target patient group includes

    potentially every person, and humans are very willing to pay for chronic medical therapy in order to

    delay the aging process. Thus, there are many convincing reasons why aging and aging-related

    diseases should be a major focus for drug discovery8. At the same time, oncology has become the

    largest therapeutic area in the pharmaceutical industry in terms of the number of projects, clinical

    trials and research and development (R&D) spending9, but despite the enormous resources being

    invested in prevention and treatment, cancer remains one of the leading causes of mortality

    worldwide.

    1.4 Selected Targets and their Involvement in Human Pathologies

    Among all possible protein targets involved in aging-related diseases and cancer, we focused our

    attention on proteasome (and its variant immunoproteasome), sirtuins and interleukin 6. These three

    targets are completely unrelated and play different roles in human cells, but the modulation of its

    activity (activation or inhibition) using small molecules could have beneficial effects on one or

    more aging-related diseases and cancer.

    Proteasome is the central catalytic unit of the ubiquitin proteasome system (UPS), which is used to

    degrade the main part of intracellular proteins. 20S standard proteasome (s-proteasome) is a

    cylinder-shaped complex that is composed of four stacked rings, each consisting of seven protein

    subunits; while associated with several regulator complexes, performs two crucial functions for cell

    metabolism: first of all, by degrading obsolete, misfolded or aberrant proteins proteasomes perform

    housekeeping function and maintain the cellular homeostasis; secondly, through the time-specific

    cleavage of short-life proteins, like transcription factors or transcription factor‟s inhibitors, are able

    to switch on/off many cellular pathways. Hence, the proteasome as central core of the UPS, is a sort

    of mandatory terminator of proteins and its inactivation leads to cellular death by apoptosis or

    necrosis. The immunoproteasome (i-proteasome) originates from the substitution of some

    constitutive catalytic subunits stimulated by pro-inflammatory citokines such as INF-γ e TNF-α,

  • 14

    and it is highly expressed in normal conditions only by specific cells types of the human body, such

    as those involved in immune-related function. Inhibiting or enhancing activity of proteasome or

    immunoproteasome could represent a promising strategy to counteract neurodegenerative diseases

    as well as cancer pathologies.

    Sirtuins are a family of NAD+-dependent enzymes that was proposed to control organismal life span

    about a decade ago. While such role of sirtuins is now debated, mounting evidence involves these

    enzymes in numerous physiological processes and disease conditions, including metabolism,

    nutritional behavior, circadian rhythm, but also inflammation and cancer. In mammals, seven

    sirtuins have been identified (SIRT1-7), of which two are predominantly nuclear, SIRT6 and

    SIRT7, two are nuclear and cytosolic, SIRT1 and SIRT2, and three are mitochondrial, SIRT3-5.

    Sirtuin activators could slow the process of cellular senescence, and therefore could be useful in

    treatment of metabolic and neurodegenerative diseases, while sirtuin inhibitors could be appealing

    for the development of new anticancer and anti-inflammatory therapies.

    Interleukin-6 (IL-6) is a pleiotropic cytokine with significant functions in the regulation of the

    immune system, and plays a pivotal role in host defense against pathogens and acute stress.

    However, increased or deregulated expression of IL-6 significantly contributes to the pathogenesis

    of various human diseases. The pathological roles of the IL-6 pathway in inflammation,

    autoimmunity, and cancer were revealed by numerous preclinical and clinical studies. Therapeutic

    strategies targeting the IL-6 pathway are in development for cancers, inflammatory and

    autoimmune diseases.

  • 15

    2. Aim of the Thesis

    The overall aim of the work presented in this thesis was the rational design of new compounds able

    to modulate activity of relevant targets involved in cancer and aging-related pathologies, namely

    proteasome and immunoproteasome, sirtuins and interleukin 6.

    The objective of the thesis was accomplished through a multidisciplinary approach that involved

    different steps:

    Step 1: Hit identification

    State-of-the-art molecular modeling techniques, mainly virtual screening methods, was applied to

    selected targets to identify a limited number of small molecules able to modulate their biological

    activity

    Step 2: In Vitro Testing

    Compounds identified during Step 1 were submitted to biological testing in vitro to measure

    biological activity and identify the structure-activity relationships (SAR) that allow understanding

    the minimum requirements for activation or inhibition of biological targets.

    Step 3: Lead Optimization

    The more promising chemical scaffolds and the SAR data coming from Step 2 were used to design

    specific structural modifications, obtained trough chemical synthesis, by introducing and modifying

    functional groups able to improve biological activity and, at the same time, to affect

    pharmacokinetic and pharmacodynamic profiles, such as selectivity towards similar targets or

    bioavailability.

    Step 4: Biological Profiling

    Lead compound obtained from Step 3 were submitted to complete biological profiling to verify the

    agreement between measured activation or inhibition of each compounds and its functional activity

    in selected tissues.

  • 16

    3. Proteasome and Immunoproteasome Inhibitors

    3.1 Proteasomes: mandatory terminators.

    Proteasome is the central catalytic unit of the ubiquitin proteasome system (UPS), which is used to

    degrade the main part of intracellular proteins. 20S standard proteasome (s-proteasome) is a

    cylinder-shaped complex that is composed of four stacked rings, each consisting of seven protein

    subunits. Each of the two inner rings contain β subunits (β1 - β7), three of which (β1, β2, β5) harbor

    the proteolytic active sites catalyzing, by their N-terminal threonine residues, a caspase-like (C-

    like), trypsin-like (T-like) and chymotrypsin-like (CT-like) activity, respectively. The α-subunits

    (α1 - α7), which compose the two outer rings, have other functions such as gating the central

    chamber (thereby enabling the entry of substrates into the inner proteolytic cavity) and the binding

    of regulator complexes like the PA700, PA28 and PA20010,11

    . The association of these regulator

    complexes leads to formation of multiple forms of proteasomes like 26S (PA700-20S) and 30S

    (PA700-20S PA700), PA28-20S and PA28-20S-PA28 complexes as well as hybrid proteasomes

    (PA28-20S-PA700). 26S/30S proteasomes recognize target proteins by the presence of

    polyubiquitin chains which are then released and the target proteins are unfolded and cleaved in a

    ATP-dependent manner12–14

    . The covalent attachment of ubiquitin to acceptor lysines in a substrate

    is a multi-step process that begins with activation of ubiquitin by E1 enzyme, which transports

    ubiquitin to an ubiquitin-conjugating enzyme (E2). The latter transfers ubiquitin to substrate either

    by itself or in cooperation with an ubiquitin ligase (E3). Afterwards, additional ubiquitins can be

    added to the first, by linkage to one of its lysines, giving rise to the polyubiquitin chain. After

    proteasome targeting, ubiquitin is recycled by deubiquitylating enzymes (DUBs), some of which

    also function to oppose the action of E3s15

    . It is worthy to note that the binding of

    polyubiquitylated proteins to the 19S regulator of mammalian and yeast 26S proteasomes enhances

    the peptidase activities of 20S proteasome about two-fold in a process requiring ATP hydrolysis16

    .

    UPS performs two crucial functions for cell metabolism: first of all, by degrading obsolete,

    misfolded or aberrant proteins proteasomes perform housekeeping function and maintain the

    cellular homeostasis; secondly, through the time-specific cleavage of short-life proteins, like

    transcription factors or transcription factor‟s inhibitors (e.g. UPS cleaves IκB-α leading to the

    entrance of NF-κB in the nucleus) proteasomes are able to switch on/off many cellular pathways.

    Hence, the proteasome as central core of the UPS, is a sort of mandatory terminator of proteins and

    its inactivation leads to cellular death by apoptosis or necrosis17–19

    .

  • 17

    Proteasome not only cleaves proteins but also can ligate two of the produced fragments, thereby

    generating peptides with a sequence that differs from the sequence of the original substrate. This

    process, also known as proteasomal splicing, has been demonstrated in vivo so far only for four

    MHC class I-restricted epitopes20–24

    , leading to the assumption that proteasomal splicing activity is

    a rare event, although recent results obtained in vitro suggest that it is in fact part of the normal

    activity of proteasomes25

    . Because of some inherent technical difficulties and the unexpected

    novelty of proteasomal splicing, the biochemical models as well as the understanding of the

    relevance of proteasomal splicing activity were so far only partially investigated. A deeper study of

    this process would have also implications from the immunological point of view, taking into

    account that PCPS highly increases the antigenic diversity26

    . Indeed, proteasomes are not only

    responsible for the degradation of the greater part of the cytoplasmic proteins but they also generate

    the vast majority of virus- or self-derived peptides presented by the MHC class I molecules on cell

    surface27,28

    . This latter function is generally aided by the interferon-γ (IFN-γ)-induced synthesis of

    the PA28-α and PA28-β proteasome activator subunits as well as of the β1i, β2i, β5i alternative

    catalytic subunits (also known as LMP2, MECL-1 and LMP7, respectively) with concomitant

    formation of the immunoproteasome (i-proteasome). The different catalytic subunits confer to i-

    proteasome differences in cleavage preferences and degradation rates, which however, vary from

    substrates to substrates29

    .

    Recently, it has been described a pivotal involvement of i-proteasome in cytokine-mediated

    inflammation in mice, because its depletion altered T cell receptor (TcR) repertoire formation, the

    number of CD8+ T cells in the spleen, T cell survival and early activation, differentiation into

    inflammatory effector cells, release of pro-inflammatory cytokines, such as IL-6 which plays a

    pivotal role also in cancer (see below), as well as response to oxidative stress30–33

    .

    Although the removal of oxidized proteins by proteasomes is clearly established34–36

    , recent works

    suggest that i-proteasomes are more prone than s-proteasome at eliminating them. Indeed, blocking

    expression of β1i by siRNA significantly reduces the adaptive response to mild oxidative stress in

    MEF cell lines37

    whereas β1i -/- mice exhibit higher levels of protein carbonyls in brain and liver

    upon aging than those of their wild-type littermates38

    . In addition, Seifert and co-workers showed

    that i-proteasome is a key element for the clearance of oxidized proteins and aggresome-like

    induced structures upon INF-γ stimuli39

    . These independent observations, gathered by exploiting

    new i-proteasome specific inhibitors as well as i-proteasome knock-out (KO) mice, opened de facto

  • 18

    a new era in the investigation of i-proteasome functions, which were so far almost merely confined

    to antigen presentation.

    It is worth to mention that between s- and i-proteasomes a group of intermediate-type proteasomes

    does exist, which are characterized by different, and tissue-specific, combinations of standard and

    inducible catalytic subunits, and probably by different post-translational modifications, able to alter

    their proteolytic activity, sensitivity to inhibitors and outer surface charge40–42

    . At last, but not least,

    the group of Tanaka described another isoform of β5 subunit (β5t) specific for thymus, which

    characterizes the so called thymus proteasome (t-proteasome), the third main isoform of

    proteasomes, having specific functions in the positive/negative selection of thymocyte and in CD8+

    T cell development43,44

    .

    3.2 Why is i-proteasome a potential therapeutic target?

    The research for specific modulators of the i-proteasomes activity is a very hot topic of today‟s

    biology for several reasons. The first one, and likely most relevant, is that i-proteasome is highly

    expressed in normal conditions only by specific cells types of the human body, such as those

    involved in immune-related function or few organs like the liver45

    , whereas the majority of cells

    barely have i-proteasome. The most famous inducer of i-proteasome synthesis is IFN- , usually

    secreted by cells during inflammation, although different studies suggested also other mechanisms,

    such as the activation of Toll-like receptor-4 (TLR-4)46,47

    . In pathological situation like

    neurodegenerative diseases, inflammatory processes induce i-proteasome synthesis in cells (e.g. in

    neurons) where normally i-proteasomes are absent48–51

    . Therefore, this disease-related expression of

    i-proteasome becomes a potential marker of pathological processes (potentially exerting both

    beneficial and detrimental effects) and possibly a therapeutic target.

    Studies carried out on animal models showed that the inhibition or the absence (in 5i KO mice, for

    example) of i-proteasomes could either ameliorate or worsen the course of the disease in a disease-

    specific manner31,32,52

    . Although the specific enhancement of the i-proteasome activity could likely

    be an appealing strategy for future therapies of selected diseases, the research in this direction is

    dampen by the absence of effective and treatment-compatible i-proteasome enhancers. Therefore,

    investigations on this topic are at present carried out only with i-proteasome inhibitors. These

    studies highlight two issues that must be carefully considered: the specificity of the inhibitors for

    the i-proteasome subunits and the potential presence of compensatory mechanisms activated in i-

    proteasome KO mice. The first issue will be discussed in next paragraph. The latter issue, on the

  • 19

    contrary, has been raised by Muchamuel et al. by suggesting that the increased amounts of 5 and

    decreased 1i and 2i subunits in 5i KO mice compared to wild type mice may mask the 5i-

    specific functions in complex cellular processes such as inflammatory responses31

    .

    3.3 I-proteasome as target for cancer therapy.

    Cancer development is a multifactorial process, which involves various genetic alterations including

    the activation of oncogenes, inactivation of tumour-suppressor genes, disregulation of cell cycle

    progression and apoptosis, as well as modification of immunosurveillance53

    . Because of the crucial

    role of proteasomes in controlling many of these biological and metabolic processes as well as the

    production of MHC class I-restricted epitopes responsible for CD8+ citotoxic T cell (CTL)

    activation54

    , this protease has become an attractive target for the treatment of malignancies55–58

    (Table 3.1). A number of preclinical studies showed that tumour cells are often more sensitive to

    proteasome blockade than normal cells and different mechanisms have been hypothesized59

    . For

    example, many types of malignant cells rapidly proliferate and might accumulate defective proteins

    at a much higher rate than normal cells, thereby increasing their dependency on proteasome as

    disposal mechanism58

    . Moreover, inhibition of proteasome causes an inactivation of NF- B

    pathway, which is involved in maintaining drug or radiation resistance in cancer cells, and it might

    reverse or bypass some alterations of cell-cycle and apoptotic checkpoint that lead to

    tumourigenesis59

    . In addition, while the activity of s-proteasome is generally found to be up

    regulated in cancer cells60

    , the levels of i-proteasome seem to vary depending on tumour types. In

    particular, i-proteasome is induced in haematological malignances, such as Multiple Myeloma

    (MM); therefore, the selective inhibition of this proteasome isoform may increase the effectiveness

    of proteasome blocking and alleviate side effects associated to non selective inhibitors of specific

    proteasome isoforms such as bortezomib, carfilzomib and NPI-005261

    (Figure 3.1 and Table 3.2). In

    accordance, first-generation i-proteasome inhibitors have been developed and tested in vitro and in

    animal model trials. Moreover, considering the involvement of proteasomes in MHC class I-

    restricted epitope production and the different contribution of s-proteasome, i-proteasome as well as

    intermediate type proteasomes to process specific antigens, the selective inhibition of these different

    isoforms could play a crucial role in CTL-based immunotherapy, as described below62

    .

  • 20

    Disease family Disease

    Type of i-

    proteasome

    modulation

    Used

    inhibitor

    Tumours MM Inhibition PR-92463

    MM Inhibition IPSI-00161

    Prostatic cancer Inhibition UK-10164,65

    Table 3.1. Pathologies where an i-proteasome specific inhibition as therapy has been tested. The list includes the main

    pathologies where an i-proteasome inhibition has been tested. MM=Multiple Myeloma.

    3.3.1 I-proteasome in Multiple Myeloma

    MM is a neoplastic plasma-cell disorder characterized by clonal proliferation of malignant plasma

    cells in the bone marrow microenvironment, accounting for approximately 1% of neoplastic

    diseases and 13% of haematologic cancers. Myeloma arises from an asymptomatic premalignant

    proliferation of monoclonal plasma cells derived from post-germinal-center B cells; multistep

    genetic and microenvironmental changes lead to the transformation into a malignant neoplasm. In

    particular, primary early chromosomal translocation occurs at the immunoglobulin switch region,

    while subsequent rearrangements, gene mutations and epigenetic dysregulation have been reported

    to affect disease progression, by altering the expression of several genes including adhesion

    molecules as well as responses to growth stimuli in the microenvironment. Interaction between

    myeloma cells and bone marrow or extracellular matrix increases tumour growth, survival and drug

    resistance, through the production of cytokines and growth factors such as IL-6, IL-10 and vascular

    endothelial growth factor among others66

    . At present, the anti-myeloma therapeutic regimes are

    based on the different combination of immunomodulatory drugs (e.g Dexamethasome,

    Lenalidomide, Prednisone) and the proteasome inhibitor bortezomib, thereby leading to the

    disruption of several signaling pathway67

    . In particular, proteasome inhibition stimulate multiple

    apoptotic pathways, including the induction of endoplasmic reticulum (ER) stress response, and by

    inhibiting the NF- B pathway, it down-regulates the productions of angiogenesis factors, cytokines

    such as IL-6 and cell adhesion molecules in the microenvironment58,66

    . However, considering the

    adverse effects related to the generalized proteasome inhibition mediated by bortezomib (e.g.

    haematological toxicity and peripheral neuropathy) and the up-regulation of i-proteasome in

    primary MM patient (CD138+) tumour cells

    63, a number of first-generation i-proteasome inhibitors

    have been tested in vitro and in animal models, showing anti-myeloma activity, mediated by several

    mechanisms. The selective inhibition of β5i subunit (e.g by PR-924 or PR-957, see below) blocks

  • 21

    growth and triggers apoptosis in MM cell lines and MM patient‟s primary cells, without affecting

    normal peripheral blood mononuclear cells. Moreover, it allows to overcome bone marrow stromal

    cells mediated drug resistances, even in presence of IL-6, which is a pro-survival factor not only for

    for MM but also for other kind of tumours, such as breast cancer68

    . Additionally, it inhibits tumour

    growth in both human plasmacytoma xenograft and SCID-hu mouse model, by decreasing the

    levels of IL-6, increasing apoptosis and inhibiting angiogenesis63

    . Noteworthy, an inhibition of the

    presentation of tumour epitopes that are β5i-dependent has also been reported31

    . The same anti-

    proliferative activity has been showed in vitro by inhibiting the 1i subunit (e.g by IPSI-001, see

    below) in lymphoid-derived tumour cells lines and MM patient-derived samples. This effect is

    exerted by the combined activation of the intrinsic and extrinsic apoptotic pathway and the

    inhibition of NF-κB signaling. Moreover, an overcome drug resistance to doxorubicin, melphalan

    and bortezomib as well as an improved toxicity profile in nonhematopoietic tissues have been

    described61

    .

    3.3.2 I-proteasome in solid tumours.

    At present, no data are available on the application of i-proteasome specific inhibitors to other kind

    of haematological malignancies such as acute myeloid leukemia (AML) and solid tumors, although

    the selective inhibition of the 1i subunit (i.e. by UK-101, see below) led to growth-inhibitory

    activity in prostatic cancer cells64,65

    . In these tumours, a heterogeneus expression of the antigen-

    processing machinery (APM), including i-proteasome subunits, is observed and often correlates to

    the progression of disease and the immune response escape53

    . Indeed, in bone marrow biopsies of

    AML patients multiple defects in APM expression were reported and remarkably a progressive

    downregulation of APM was seen from initial diagnosis to relapse69

    . In renal carcinoma cells lower

    levels of 1i and 5i subunits as well as of the epitope transporters into ER (i.e. TAPs) have been

    found and they were more pronounced in metastatic lesions than primary tumour53

    . The same

    scenario has been observed in hepatocellular carcinoma cell lines70

    and primary malignant

    melanoma lesions, associated with lack of spontaneous regression71

    whereas mice lacking 1i

    subunit develop spontaneous uterine leiomyosarcoma72

    . An up-regulation of i-proteasome and an

    increased CTL response against tumour antigens have been observed in vitro in hepatocarcinoma

    cell lines after the administration of INF-70

    , suggesting that in certain cases restoration rather than

    inhibition of i-proteasome functionality could be effective in anti-tumour induced-response, by

    enhancing the production of tumour-specific MHC class I-restricted epitopes.

  • 22

  • 23

  • 24

    Figure 3.1. Chemical structures of promiscuous proteasome inhibitors.

  • 25

    Structure Selectivity Inhibition

    mechanism Subunit Activity

    1 All All reversible

    2 β5, β5i CT-like irreversible

    3 β2, β5, β5i, β2i CT-like>T-like>C-like irreversible

    4 β5, β5i CT-like irreversible

    5 β5, β1i CT-like reversible

    6 All All irreversible

    7 β1, β1i C-like irreversible

    8 β2 T-like irreversible

    9 β5, β5i CT-like irreversible

    10 β5 CT-like reversible

    11 β1, β2, β5 CT-like>T-like>C-like irreversible

    12 β5 CT-like reversible

    13 β5 CT-like>T-like, C-like reversible

    14 β2>β5>β1 T-like>CT-like>C-like irreversible

    15 β2>β5>β1 T-like>CT-like>C-like irreversible

    16 β5>β1, β2 CT-like>C-like, T-like irreversible

    17 β5>β1, β2 CT-like>C-like, T-like irreversible

    18 All All irreversible

    19 β5 and β2 CT-like>T-like irreversible

    20 β5>β1,β2 CT-like>C-like, T-like reversible

    21 β5 CT-like reversible

    22 β5 CT-like non covalent

    23 β5 CT-like reversible

    24 β5>β2, β1 CT-like> T-like, C-like non covalent

    25 β5 CT-like

    26 β5 CT-like

    27 β1 / β5 C-like / CT-like irreversible

    28 β5 CT-like non covalent

    Table 3.2. Activity profiles of promiscuous inhibitors of s- and i-proteasomes. Structure enumeration of proteasome

    inhibitors refers to what reported in Fig. 3.1. Proteasome activities, as defined with short-fluorogenic substrate assay,

    are shortened as following: CT-like = chymotrypsin-like, T-like = trypsin-like, C-like = caspase-like.

  • 26

    3.3.3 I-proteasome as a target for immunotherapy?

    The concept of using vaccination in the treatment of cancer has been tied to the history of vaccines

    themselves. However, the major interest in it has recently grown with the increasing understanding

    of the role that the immune system (IS) plays in shaping the biological behavior of cancer, including

    the identification of tumour antigens recognized by CTLs73

    . As above mentioned, the MHC class I-

    restricted antigen presentation is generally enhanced after IFN- stimuli by inducing the expression

    of i-proteasomes, PA28- , TAPs and MHC class I molecules29

    . It has been proposed that i-

    proteasome increases the production of MHC class I-restricted epitopes because of its higher

    inclination for generating peptides with hydrophobic and basic C-termini, which shall have a better

    affinity for the APM74

    . Nevertheless, the group of Van den Eynde reported some examples of MHC

    class I-restricted tumour (self)-epitopes that were better generated by s-proteasome than i-

    proteasome75–78

    . They speculated that s-proteasome could better generate self-epitopes in contrast to

    i-proteasome more efficient in viral epitope production75

    . This difference between s- and i-

    proteasome might have fallouts on CD8+ T cell-mediated immune response at different levels,

    including thymocytes development, tolerance induction, CTL activation as well as cancer

    immunotherapy. Although this theory is quite appealing, we believe that the limited number of self-

    and viral-epitopes properly investigated so far does not allow to statistically confirm it. In addition,

    this theory recently evolved because the group of Van den Eynde showed also that two tumour

    epitopes of MAGE-A*03 and –A*10 proteins are processed exclusively by intermediate type

    proteasomes, which are variabily expressed in tumour, dendritic cells and in normal tissues18,79,80

    .

    In summary, the results obtained in the last decade suggest that the repertoire of antigens presented

    by a given cell is strongly affected by its proteasome content, composition as well as the levels of

    their regulators, which could modify cleavage properties of protein substrates81

    . This observation

    could have strong implications on cancer immunosurveillance although further investigations are

    mandatory to define the role of different protesome isoforms in immunotherapy against cancer.

    3.4 I-proteasome as target for neuropathologies

    Although in young healthy human central nervous system (CNS) i-proteasomes are almost absent,

    they have been detected in cells of different CNS areas from elderly subjects as well as from

    patients affected by Alzheimer (AD) or Huntington (HD) diseases49,51

    , Multiple Sclerosis (MS) with

    a concomitant expression of the PA28- complex50

    and very recently in Temporal Lobe Epilepsies

    (TLE)48

    (Table 3.1). The induction of i-proteasome and PA28- expression in brain could have

  • 27

    effects on different pathways because UPS regulates in neurons, for example, the pre- and post-

    synaptic plasticity and the protein turnover82

    . Processes and mechanisms responsible of cerebral i-

    proteasome synthesis are still unknown. Neuroinflammation could be the trigger of i-proteasome

    expression as we hypothesized for aging, AD, MS and TLE48–50

    , which will be briefly here

    discussed as example of age-related (i.e. AD) or autoimmune (i.e. MS) neurodegenerative diseases

    and epilepsies (i.e. TLE). It is worthy to note that i-proteasome synthesis induction could be limited

    to CNS or be the result of a phenomenon started in periphery and transferred to CNS, as it might

    occur during the onset of MS. Accordingly, IS regulation in other organs could have implications

    also at CNS level, including the regulation of i-proteasome expression. For example, a cross-talk

    between brain and gut with implications for the IS has been proposed recently in studies on

    experimental autoimmune encephalomyelitis (EAE)83–85

    .

    3.4.1 Alzheimer disease.

    AD is a devastating neurodegenerative disorder of the CNS occurring most frequently in later stages

    of adulthood. AD is associated with a specific pattern of pathological changes in brain that result in

    neurodegeneration and progressive development of dementia. These pathological hallmarks of AD

    are neuronal loss accompanied by intraneuronal neurofibrillary tangles formed of tau-based paired

    helical filaments (PHFs) and extracellular senile plaques of -amyloid86

    . It has been reported that

    PHFs inhibit proteasome activity and it has been suggested that this inhibition may induce neuronal

    damage in AD87

    . Furthermore, UPS is also involved in the control of the physiological maturation

    of the -amyloid precursor protein by modulating the intracellular concentration of presenilins88

    .

    An inhibition of proteasome activity in crude extracts from AD affected brain areas has been

    described, whereas an investigation carried out on 20S proteasome purified from the frontal

    neocortex of AD patients suggested that the observed drop of 20S proteasome activity in AD tissue

    could be due to the presence of inhibitory molecules more than to an intrinsic decrease of the 20S

    proteasome functionality87,89,90

    .

    AD occurs usually in elderly brain, where i-proteasome expression is already present in different

    cells types, maybe as a result of the inflamm-aging phenomenon and/or as attempt to cope with the

    increasing oxidative stress91

    . I-proteasome expression is further induced in hippocampi (but not in

    cerebellum) of AD patients49

    and its presence varies among different neuronal sub-types. It has

    been speculated that i-proteasome expression might be an attempt to tackle the accumulation of

    oxidised proteins and phosphorylated tau that occur during AD progression, since they are both

    preferentially degraded by i-proteasome39,92

    . However, the few available information about the role

  • 28

    of i-proteasome in AD does not allow us to estimate if an inhibition of i-proteasome function would

    lead to a progression or a reduction of the neurological damage. In addition, AD clinical symptoms

    emerge when the neurological damage is already pronounced; therefore, we might speculate that a

    modulation of i-proteasome function at the early stages of the disease could have different effects

    (even opposite) than a modulation in the late stages of AD.

    3.4.2 Multiple Sclerosis.

    MS is the most common autoimmune disorder of the CNS. It is characterized by multifocal areas of

    demyelization (plaques), chronic inflammation and damage to oligodendrocytes and neurons. The

    cause of MS is still unknown and disease pathways are poorly understood. However, the association

    of HLA-DRB1*15 and other HLA class I (e.g. HLA-A*02 and HLA-A*03) and class II alleles, the

    presence of autoreactive T lymphocytes together with other inflammatory cells and cytokines in

    active MS lesions suggest an autoimmune pathogenesis. Accordingly, EAE, a classical mouse

    model for MS, can be induced by the administration of myelin antigens or CD4+ and CD8

    + T

    lymphocytes specific for those antigens93–95

    . It has been proposed that the first bout of the disease is

    mediated by CD8+ T cells while the first relapse and MS progression are mediated by CD4

    + T cells

    through different mechanisms such as antigen release and epitope spreading96

    .

    Although preliminary observations on white and grey matters of MS patients suggested that 20S

    and 26S proteasomes activity is decreased compared to controls97

    , no information are so far

    available on proteasome activity in plaques, although an accumulation of i-proteasome has been

    observed50

    . In CNS of subjects affected by MS, i-proteasomes are expressed in different cell types

    such as oligodendrocytes, astrocytes, macrophages/microglia, infiltrating lymphocytes and, weakly,

    in neurons as demonstrated by double IHC assays. A similar expression has been described also for

    the subunit of the PA28 complex. Intriguingly, the polymorphic variant HH at the codon 60 of

    1i subunit was significantly less present in a sizeable Italian MS population (OR = 0.44) restricted

    to females carrying the HLA-A*02 allele50

    . The Authors correlated this genetic data to the

    observation that a HLA-A*02-restricted epitope (MBP111-119), which activates memory T cells

    preferentially in blood of MS patients98–100

    , was produced in lower amount during in vitro digestion

    by 20S i-proteasome carrying the variant HH at the codon 60 of 1i subunit. Authors speculated

    that a lower production and presentation of this epitope as well as other myelin epitopes bound to

    the HLA-A*02, could reduce the probability to disrupt the physiological tolerance (central or

    peripheral) of myelin-specific CTLs and/or their cytotoxicity towards oligodendrocytes, thereby

    restraining the MS onset50

    . The i-proteasome activity and the 1i R60H polymorphism might have

  • 29

    implications also at thymic level during the selection of myelin –reactive thymocytes although no

    exhaustive information is available to this regard.

    Furthermore, the study of Seifert and colleagues suggested another i-proteasome-related mechanism

    that could affect the MS onset. Indeed, they showed that i-proteasome is essential for an efficient

    clearance of oxidised and polyubiquitylated proteins upon IFN- induced oxidative stress, thereby

    preserving protein homeostasis during inflammation. Accordingly to this observation, they reported

    that 5i ko mice showed an earlier onset and worst clinical score than wild type mice in an EAE

    model39

    . Although these two studies would suggest that i-proteasomes may influence onset and

    progression of MS affecting both the myelin-specific CD8+ T cell activity and the response of

    different cell types to the inflammatory aggression both in periphery and in the CNS, further

    independent confirmations are needed before drawing a model connecting i-proteasome and MS

    which could have a remarkable spin-off for future therapeutic approaches to the disease.

    3.4.3 Temporal Lobe Epilepsy.

    Epilepsy is a neurological disorder that affects about 50 million people worldwide and is

    characterized by an enduring predisposition to generate seizures as well as by emotional and

    cognitive dysfunctions. About 30% of epileptic patients are defined pharmacoresistant since they do

    not adequately respond to therapies and in these patients, affected by TLE, the surgical removal of

    the epileptic focus is often the only therapeutic option to achieve seizure control101

    .

    The evidences available from experimental and clinical findings support a crucial role of immune

    and inflammatory processes in the aetiopathogenesis of epilepsy47

    . Pronounced inflammatory

    processes have been described in human epileptogenic brain tissue from TLE and epilepsies

    associated with malformations of cortical development, where seizures are often refractory to

    anticonvulsant treatments102

    .

    Pharmacological studies in experimental models and the use of transgenic mice with perturbed

    cytokine systems showed that proinflammatory cytokines (e.g. IL-1 , TNF- ), danger signals (e.g.

    HMGB1), complement factors and prostaglandins significantly contribute to seizure activity and

    cell loss, and that inhibition of the production of these molecules or blockade of their receptors (e.g.

    ILR1, TLR-4), significantly reduced seizure activity103

    .

    Recently, i-proteasomes have been detected in cortex and hippocampus of patients affected by

    different TLE forms48

    . By IHC staining i-proteasomes were revealed in glia and neurons of TLE

    hippocampi whereas controls showed positivity to 1i and 5i only in luminal endothelial cells, as

  • 30

    observed in other studies49,50

    . Intriguingly, the neuronal i-proteasome expression differs between

    TLE forms. Furthermore, no IFN- was detected in TLE specimens suggesting that in this disease i-

    proteasome synthesis is induced by a different mechanism. A good candidate could be TLR-4,

    which can be triggered by LPS treatment, a well-known i-proteasome inducer. Thus, taking into

    account the role that TLR-4 plays in the disease, we might speculate that i-proteasome is one of the

    molecules induced/activated by TLR-4 pathway that mediate the effects that this receptor has

    during epileptogenesis. Further investigations are however mandatory to address this issue as well

    as to understand the role of i-proteasome in the different neuroinflammatory processes involved in

    epilepsy.

    It is worth to note that so far no studies have exploited the availability of i-proteasome ko mice by

    crossing them with disease animal models (e.g. APP/PS1 model for AD) or inducing a disease-

    associated symptoms by drugs such as pilocarpine or kainite (as epilepsy models), respectively. We

    surmise that these types of studies will provide breakthrough information on the disease

    aetiology/progression and the involvement of i-proteasome in the pathological mechanisms.

    3.5 An overview of selective s- and i-proteasome inhibitors and

    enhancers.

    Despite the efforts made in recent years to discover selective inhibitors for either s-proteasome or i-

    proteasome, only six compounds showing some selectivity (two for the s-proteasome, PR-893 and

    PR-825, and four for the i-proteasome, PR-924, PR-957, IPSI-001 and UK-101) have been

    described (Table 3.2 and Figure 3.2). PR-893 (compound 1 in Fig. 3.2) is a tripeptide epoxyketone

    which shows a selectivity of 20 folds for β5 over β5i subunits104

    . This molecule was used by Parlati

    and co-workers to prove, in their pioneering study, the relationship between the selective inhibition

    of subunits (β5 and β5i) with chymotrypsin-like (CT-like) activity and the antitumour response in

    tumour cells of haematological origin. The same reactive portion of PR-893 can be found in PR-825

    (compound 2 in Fig. 3.2), an analogue of carfilzomib (compound 2 in Fig. 3.1), which has been

    synthesized by Zhou et al. by varying the P2, P3 and N-Cap of bortezomib105

    . These variations gave

    rise to an inhibitor of proteasomal CT-like activity, which is about 14 folds more selective towards

    β5 as respect to β5i. On the other hand, two selective inhibitors related to carfilzomib (compound 2,

    in Fig.3.1), namely PR-924 (comp. 3, Fig. 3.2) and PR-957 (comp. 4, Fig. 3.2), have been

    developed to target β5i subunit of the i-proteasome106

    . As in the case of the above-mentioned s-

    proteasome inhibitors, these two compounds are tripeptide epoxyketones. Their structure differs

  • 31

    from s-proteasome selective inhibitors for the presence of three marked aromatic and hydrophobic

    pharmacophoric features. In particular, both PR-924 and PR-957 have two aromatic moieties in the

    proximity of the reactive group, i.e. epoxyketones phenylalanine instead of epoxyketone leucine of

    PR-893 and PR-825 as well as bulkier tryptophan (PR-924) and tyrosine (PR-957) instead of

    smaller side-chain amino acids for PR-893 and PR-825. Equally, the N-protecting groups are also

    more hydrophobic and large as respect to s-proteasome inhibitors. All these hydrophobic/aromatic

    features are expected to interact with specific moieties of the β5i binding site and, as discussed

    below, these may constitute the molecular reason for the i-proteasome selectivity over s-proteasome

    catalytic activity. In fact, PR-957 is reported to be 20- to 40- more selective for murine β5i over β5

    and β1i subunits and, at the concentration which allows minimal impact on other subunits, it

    inhibited the presentation of β5i-dependent epitopes31

    . Equally, PR-924 was shown to be 100-fold

    more selective for human β5i and less selective for CT-like activity of β5 as compared to

    bortezomib and carfilzomib63

    . The selective inhibition of i-proteasome has been obtained also with

    a dipeptide aldehydes compound, IPSI-001 (compound 5, Fig. 3.2) that targets the β1i subunit and

    inhibit i-proteasome C-like and CT-like activity in vitro in lymphoid-derived tumour cells lines and

    MM patient-derived samples. Similarly to PR-924 and PR-957, IPSI-001 has three hydrophobic and

    extended features such as the carboxybenzyl N-protecting group and the n-propyl side chain close

    to the reactive aldehyde. Finally, another β1i specific inhibitor, UK-101, (compound 6, Fig. 3.2) has

    been developed by Ho and co-workers, demonstrating growth-inhibitory activity in prostatic cancer

    cells expressing higher level of β1i with no effect on s-proteasome function64,65

    . UK-101, like IPSI-

    001, is also a dipeptide with a tert-butyldimethylsilyl group attached at the C-terminal hydroxyl

    group65

    . All the above-mentioned s- and i-proteasome inhibitors covalently modify the catalytic N-

    terminal Thr of the proteolytic β subunits thereby affecting specific activities by means of covalent

    reversible or irreversible inhibition mechanisms (Table 3.3). Considering the large number of

    promiscuous inhibitors of s- and i-proteasome, which can bind reversibly or irreversibly the

    catalytic Thr, such as epoxyketones, aldehydes, vinylsulfones, β-lactones, boronic acids,

    isocoumarins and Michael acceptors (Fig. 3.1 and Table 3.2), it should be pointed out that only

    epoxyketones and aldehydes have been reported as i- and s-proteasome specific inhibitors

    (compounds 1-6 in Fig. 3.2 and Table 3.3). In this regard, it should be noted that, while some efforts

    have been done to modify the reactive threonine-trap of s-proteasome inhibitors107,108

    , no similar

    strategies have been yet explored to modify known specific i-proteasome scaffolds with other

    reactive groups. It is therefore unclear whether other Thr-traps may be conveniently used for the

    design of novel i-proteasome specific inhibitors. Similarly, it is interesting to note also that non-

  • 32

    peptide-like compound such as Salinosporamide (compound 11 in Fig. 3.1), Belactosin (compound

    16-17 in Fig. 3.1) and Betulinic acid derivatives (compound 22 in Fig. 3.1) have been so far only

    described among promiscuous s- and i-proteasome inhibitors (Fig. 3.1). Even in this case, it is still

    to be ascertained whether it is possible to optimize classes of non-peptide-like compounds in order

    to reach some selectivity either for s- or i-proteasome. In this context, new insights on

    immunoproteasome structure obtained either by modelling results (see next paragraph) or new

    crystallographic resolutions promise to be highly relevant for the rational design of novel selective

    inhibitors. Although the inhibition mechanism of proteasomes has been extensively reviewed by

    several authors106,109–112

    , few information about mechanisms of activation or reversible inhibition

    are available. For example, betulinic acid derivatives (compound 22 in Fig. 3.1) were proved to

    inhibit CT-like activity113

    but there is no specific information about their inhibition mechanisms, as

    well as for Argyirn A (compound 24 in Fig. 3.1)114

    . On the other hand, Gallastegui et al.

    demonstrated, by resolving the crystal structure of s-proteasome in complex with the most potent

    hydroxyurea derivative (PDB ID: 3SHJ)115

    , that new hydroxyurea derivatives (compound 28 in Fig.

    3.1) bind non-covalently the site with CT-like activity. In contrast to the development of 20S

    inhibitors, drug-like molecules that can activate or enhance proteasome activity are, at present, rare

    and not well characterized, even if they could represent an appealing strategy for specific diseases

    in which proteasome activity has to be increased to cure the patient. Apart the role of PA28, PA700

    and PA200 regulators in proteasome activation, several type of small molecules including SDS,

    lipids (oleic and linoleic acids) and peptide–based compounds were shown to activate proteasome at

    relatively high concentrations116

    . On the contrary, oleuropein (compound 8, Fig. 3.2),

    Phaeodactylum tricornutum algae extract and betulinic acid (compound 7, Fig. 3.2) can activate

    proteasome at low micromolar concentration. Oleuropein is the major component isolated from the

    Olea europaea, it is able to increase in vitro all three proteolytic activities and delay replicative

    senescence of human embryonic fibroblast117

    . The same results have been obtained by

    Phaeodactylum tricornutum algae extract, which stimulates all three proteolytic activities of

    proteasome in vitro and in human keratinocytes thereby reducing the level of oxidized proteins118

    .

    Conversely, betulinic acid, a triterpene derived from many plant species, preferentially activates the

    CT-like activity of proteasome119

    , even if inhibitory effects have also been reported as above

    described. Finally, Chondrogianni and co-workers identified quercetin (compound 9, Fig. 3.2) and

    its derivative, namely quercetin caprylate (QU-CAP) as a proteasome activator with anti-oxidant

    properties that can influence cellular lifespan, survival and viability of HFL-1 primary human

    fibroblasts. Moreover, when these compounds were supplemented to already senescent fibroblasts,

  • 33

    a rejuvenating effect was observed120

    . The mechanism of action of all these compounds is supposed

    to be linked to conformational changes of the channel gates regulated by the subunits of the

    proteasome although further investigations are needed to characterize their enhancer properties. In

    addition, recent and ongoing research aim to elucidate the roles of other components of the UPS has

    identified several enzymes, beside the 20S catalytic core, that can be additional targets for

    therapeutic intervention by small-molecule modulators121

    . In particular, an enhancement of

    proteasome activity by a small-molecule inhibitor of the DUB USP14 has been reported122

    , as well

    as an anticancer activity of another compound which inhibits both UCHL5 and USP14 enzymes123

    ,

    suggesting that the deubiquitinating activity of the PA700 regulator could represent a new

    anticancer drug targets.

    3.6 Human immunoproteasome model.

    Despite the importance of i-proteasome as an emerging biological target for cancer and

    neuropathologies, until 2013 no crystallographic structure of the human form has been solved. A

    possible strategy to compensate that lack of knowledge was the generation of in silico human i-

    proteasome models, which could provide helpful hints for the development of selective i-

    proteasome modulators. Accordingly, we generated the human model of the whole i-proteasome by

    starting from the X-ray structure of the mammalian 20S proteasome (PDB id: 1IRU) and

    substituting of the six catalytic subunits of the inner rings with the related i-proteasome subunits

    (Table 3.4).

    At first we checked the similarity between subunits in the two inner rings of the proteasome

    structure. Root mean square deviation (RMSD) for each couple of homolog-subunits was found to

    be in the range 0.6-1.3 on all atoms, thereby indicating that homolog- -subunits of the two inner

    rings can be considered equivalent. The raw models of i-subunits were taken from the

    SwissModel repository124

    and aligned to chain H, I, L for 1, 2 and 5, respectively. The same

    models were used also to represent the i-proteasome subunits of the other inner ring. The entire i-

    proteasome model was generated by substitution of the six catalytic chains and followed by several

    structural corrections/refinements such as hydrogen addition, water and ions removal, manual

    charges corrections, addition of missing side chains and optimization of hydrogen bonds. In order to

    analyze at the molecular level the aminoacidic differences between s- and i-proteasome we reported

    in Fig. 3.3 and Table 5.3 a pair-wise comparison of the subunit catalytic pockets of the

  • 34

    mammalian s-proteasome crystallographic structure (PDB id: 1IRU) and the model of human i-

    proteasome that we derived.

    Figure 3.2. Chemical structures of selective s- and i-proteasome inhibitors and activators of proteasomes.

  • 35

    Compound

    Selectivity Mechanism and

    biological effect Subunit Activity

    1 β5 CT-like irreversible inhibitor

    2 β5 CT-like irreversible inhibitor

    3 β5i CT-like irreversible inhibitor

    4 β5i CT-like irreversible inhibitor

    5 β1i

    CT-like > C-like > T-

    like

    BrAAP

    reversible inhibitor

    6 β1i CT-like irreversible inhibitor

    7 αa activator

    8 αa activator

    9 αa activator

    Table 3.3. Activity profiles of s- and i-proteasome inhibitors as well as proteasome enhancers. Inhibition form,

    inhibited proteasome activities and relative references are reported. Proteasome activities, as defined with short-

    fluorogenic substrate assay, are shortened as following: CT-like = chymotrypsin-like, T-like = trypsin-like, C-like =

    caspase-like. Structure enumeration of proteasome inhibitors refers to what reported in Fig. 3.2. αa: the mechanism of

    these compounds has been hypothesised to be related to conformational change perturbations of α-type subunits.

  • 36

    Figure 3.3. Comparison of s-proteasome and i-proteasome β-subunits in complex with bortezomib. S-proteasome

    subunits were taken from the crystallographic structure of the human 20S proteasome (PDB id: 1IRU) while i-

    proteasome subunits were derived by our model (see above). Initial binding poses of bortezomib (compound 1 in Fig.

    1) were taken by the crystal structure of the yeast proteasome (PDB id: 2F16) aligned with correspondent subunits of

    the human proteasome (PDB id: 1IRU) and followed by ligand minimizations. Contour maps were generated with the

    software SiteMap [136], thereby producing hydrophobic (yellow regions), donor (blue regions) and acceptor (red

    regions) potentials. Contour maps represent the ideal region of the space where a corresponding ligand feature should

    be located in order to interact optimally with the subunits.

  • 37

    The catalytic binding sites of s- and i-proteasome generally show a similarity in shape and volume

    between β1, β2 and β5 and their corresponding β1i, β2i and β5i subunits. This simple fact might

    suggest that ligands with similar shape and volume properties fit equally in s- and i-proteasome

    homologue subunits. However, a deeper structural analysis shows that non-negligible differences

    between s-proteasome and i-proteasome are present in the aminoacidic composition of some of the

    catalytic pocket residues. In particular, we analyzed residues of the binding site that differ from s- to

    i-proteasome chains observing that β1 and β1i subunits show the higher degree of variability since

    ten residues differ in the substrate binding site (Fig. 3.3 and Table 5.3). On the other hand, five

    residues differ from β5 and β5i subunits whereas only four residues change from β2 to β2i subunits.

    Most interesting is to analyze the differential nature and position of these changes vis-à-vis to the

    binding of putative s- or i-proteasome inhibitors. We did such analysis by taking advantage of the

    structural information available from co-crystallized structure of bortezomib with yeast s-

    proteasome (PDB id: 2F16). In Fig. 3.3 we graphically reported the location of the aminoacids

    listed in Table 5.3 and depicted binding site differences in terms of contour maps that represent the

    ideal region of the space where a ligand feature should be located in order to interact optimally with

    the single subunits. In some cases differences involve relevant changes of aminoacid properties that

    are likely to be important for the binding of putative selective inhibitors. For instance Arg45 of the

    β1 subunit is substituted to a Leu45 in the β1i subunit. This change implies the existence of a

    noticeable hydrophobic region in the binding site of the β1i subunit (yellow region, Fig. 3.3) that is

    not present in the β1 subunit due to the polar nature of the arginine residue. Despite such a

    difference occurs in the binding site of both subunits, bortezomib seems not to be influenced by this

    change and this might constitute one of the reasons behind the promiscuous nature of this inhibitor

    towards s- and i-proteasome. Another major difference involves residues of Ala27 and Ser28 of the

    β5 subunit, which are inversed in the β5i subunit, i.e. Ser27 and Ala28. This simple inversion

    relocates the hydrophilicity of the serine residue thereby becoming more accessible for the putative

    binding of ligands. Even in this case the non-selectivity of bortezomib may be explained by the fact

    that its binding is not influenced by such aminoacidic switch, although selective lead compounds

    might be developed in light of these considerations. In contrast, β2 and β2i subunits seem to be very

    similar in terms of aminoacidic properties. This fact is graphically reflected by similar shapes and

    colours of the contour maps of Fig. 3.3. Thus, from these data, it appears particularly challenging to

    exploit differential binding site composition of β2 and β2i subunits in order to conceive selective

    inhibitors. This consideration may also explain why no selective s- or i-proteasome inhibitors

    targeting the trypsin-like (T-like) activity have been discovered so far. Finally, by comparing β1, β2

  • 38

    and β5 subunits it is interesting to note that both in the crystal structure of the yeast proteasome

    (PDB id: 2F16) as well as in our i-proteasome model, bortezomib assumes a markedly different

    molecular conformation in β2 and β2i while other subunits, i.e. β1/β1i and β5/β5i seem to

    accommodate the ligand with the same binding mode (Fig. 3.3). Huber et al.125

    published several

    crystal structures of the yeast 20S proteasome and of the mouse 20S s-proteasome and i-proteasome

    in presence or absence of the i-proteasome specific inhibitor PR-957 (compound 4 in Fig. 3.2; PDB

    ids: 3UN4, 3UN8, 3UNB, 3UNE, 3UNH and 3UNF). Through the analysis of the crystallographic

    structure the authors identified a unique catalytic feature for the i-proteasome β5i active site and,

    together with conformational changes occurring upon ligand binding, could rationalize the

    selectivity of PR-957 towards the β5i subunit. Importantly, the superposition of the above-described

    crystallographic subunits β1i, β2i and β5i with those obtained by our human i-proteasome model

    shows a striking consistency, thereby underlining the importance to obtain structural and/or

    modelling data as an effective tool for the identification of potential small-molecule lead structure,

    as described below.

    3.7 Computer-aided drug design approaches

    Few approaches of computational drug design have been applied in the last years for the discovery

    of new lead compounds able to inhibit s-proteasome126–128

    . For instance, Reboud-Ravaux and

    colleagues carried out a multistep structure-based virtual ligand screening strategy and were able to

    identify several novel lead compounds inhibiting s-proteasome with micromolar range activity and

    reported cytotoxicity on human tumour cell lines127,129

    . It is worth noting that no computer-aided

    drug design techniques have been yet reported for the discovery of selective i-proteasome

    inhibitors. In this context, likely, new insights on i-proteasome structure obtained by computational

    modelling128

    , including the present work, or by means of new crystallographic evidences125

    , might

    constitute a new way to deploy chemoinformatic techniques such as docking or pharmacophore

    high-throughput virtual screenings130,131

    on large database of chemicals compounds132,133

    . In

    particular, it appears that differences of aminoacids (Fig. 3.3 and Table 5.3) in the s- and i-

    proteasome β subunit binding sites (especially β1 versus β1i and β5 versus β5i) might effectively

    constitute the molecular basis for the development of novel s- and i-proteasome specific inhibitors,

    keeping however into account that the design of highly-specific inhibitors may still result in

    challenging tasks because of the major similarities in the β subunit catalytic sites.

  • 39

    Proteasome

    subunit – gene

    name

    Common

    Name Uniprot code

    Chain name

    (PDB id:

    1IRU)

    Immuno-

    proteasome

    subunit – gene

    name

    Common

    Name Uniprot code

    PSMB6 1 P28072 H/V PSMB9 1i P28065

    PSMB7 2 Q99436 I/W PSMB10 2i P40306

    PSMB5 5 P28074 L/Z PSMB8 5i P28062

    Table 3.4. S- and i-proteasome catalytic subunit nomenclature.

    Subunits Proximal residues (within 5Å from bortezomib pose)

    20 21 22 23 27 28 31 45 46 48 52 95 97 116

    β1 T T T A T R T M G M

    β1i V S A V F L A S H G

    β2 E G T T

    β2i N D V A

    β5 T A S A G

    β5i S S A S C

    Table 3.5. Proximal amino acidic differences in the catalytic binding sites of s- and i-proteasome β-subunits.

    3.8 Hit Identification

    Since the growing amount literature data suggesting the pivotal role of i-proteasome in different

    cellular pathways, including apoptosis and inflammation, a selective modulation of i-proteasome,

    by inhibiting or enhancing its activity, directly with small-molecule modulators or indirectly, e.g. by

    regulating E3 and DUB enzymes, could represent a promising strategy to counteract these

    pathologies. At the time of this study, medicinal chemistry efforts identified a small number of low

    molecular-weight inhibitors that are able to modify 20S s- and i-proteasome functions, but only few

    examples of selective i-proteasome inhibitors was developed. So we started an hit identification

  • 40

    campaign to find new selective i-proteasome inhibitors using virtual screening methodologies. The

    available x-ray structure of human proteasome (PDB code 1IRU) and the generated model of i-

    proteasome, after a standard protein preparation procedure, were used as template to virtually